1
|
Pousa S, Ramos-Bermúdez PE, Besada V, Cabrales-Rico A, Guirola Cruz O, Garay HE, Rodríguez-Mallón A, Zettl K, Wiśniewski JR, González LJ. Characterization by LC-MS/MS analysis of KLH vaccine conjugated with a tick antigen peptide. Analyst 2025; 150:1091-1102. [PMID: 39817672 DOI: 10.1039/d4an01449a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Keyhole limpet haemocyanins (KLH1 and KLH2) from Megathura crenulata, are multi-subunit oxygen-carrying metalloproteins of approximately 3900 amino acids, that are widely used as carrier proteins in conjugate vaccines and in immunotherapy. KLHs and their derived conjugate vaccines are poorly characterized by LC-MS/MS due to their very stable supramolecular structures with megadalton molecular mass, and their resistance to efficient digestion with standard protocols. KLH1 and KLH2 proteins were conjugated to the conserved P0 peptide (pP0), derived from the P0 acidic ribosomal protein of Rhipicephalus sp. ticks using maleimide-thiol chemistry to obtain a broad-spectrum anti-tick vaccine. The resulting KLH1- and KLH2-Cys1pP0 conjugate vaccines were efficiently digested using the Multiple-Enzymatic Digestion Filter Aided Sample Preparation and analyzed by LC-MS/MS, enabling a sequence coverage of approximately 85% of both conjugates. Seventy-three and sixty-five percent of all lysine residues in KLH1 and KLH2, respectively, were partially conjugated to Cys1pP0. In the quaternary structures, we found no bias toward conjugation of lysine residues exposed to either the outer surface or the inner channel. The latter may not contribute to a protective humoral response because B cell entry into the inner channel is incompatible with the entrance hole diameter. The Cys-His thioether bonds in both KLHs were determined by identifying type 1 cross-linked peptides. New post-translational modifications undescribed for the KLH such as oxidized species, were identified. This is the first report of the identification of conjugation sites of two KLH-based vaccines. These results will help translate the KLH-based conjugates into well-characterized biotechnology products.
Collapse
Affiliation(s)
- Satomy Pousa
- Department of Proteomics, Mass Spectrometry Laboratory, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba.
| | - Pablo E Ramos-Bermúdez
- Bioinformatics, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba
| | - Vladimir Besada
- Department of Proteomics, Mass Spectrometry Laboratory, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba.
| | - Ania Cabrales-Rico
- Purification and Analytic Group, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba
| | - Osmany Guirola Cruz
- Bioinformatics, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba
| | - Hilda Elisa Garay
- Laboratory of Peptide Synthesis, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba
| | - Alina Rodríguez-Mallón
- Animal Biotechnology, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba
| | - Katharina Zettl
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Munich, Germany
| | - Jacek R Wiśniewski
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Munich, Germany
| | - Luis Javier González
- Department of Proteomics, Mass Spectrometry Laboratory, Center for Genetic Engineering and Biotechnology, 31 Avenue, Cubanacan, Playa, Havana, Cuba.
| |
Collapse
|
2
|
Hu J, Nieminen AL, Zhong Z, Lemasters JJ. Role of Mitochondrial Iron Uptake in Acetaminophen Hepatotoxicity. LIVERS 2024; 4:333-351. [PMID: 39554796 PMCID: PMC11567147 DOI: 10.3390/livers4030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Overdose of acetaminophen (APAP) produces fulminant hepatic necrosis. The underlying mechanism of APAP hepatotoxicity involves mitochondrial dysfunction, including mitochondrial oxidant stress and the onset of mitochondrial permeability transition (MPT). Reactive oxygen species (ROS) play an important role in APAP-induced hepatotoxicity, and iron is a critical catalyst for ROS formation. This review summarizes the role of mitochondrial ROS formation in APAP hepatotoxicity and further focuses on the role of iron. Normally, hepatocytes take up Fe3+-transferrin bound to transferrin receptors via endocytosis. Concentrated into lysosomes, the controlled release of iron is required for the mitochondrial biosynthesis of heme and non-heme iron-sulfur clusters. After APAP overdose, the toxic metabolite, NAPQI, damages lysosomes, causing excess iron release and the mitochondrial uptake of Fe2+ by the mitochondrial calcium uniporter (MCU). NAPQI also inhibits mitochondrial respiration to promote ROS formation, including H2O2, with which Fe2+ reacts to form highly reactive •OH through the Fenton reaction. •OH, in turn, causes lipid peroxidation, the formation of toxic aldehydes, induction of the MPT, and ultimately, cell death. Fe2+ also facilitates protein nitration. Targeting pathways of mitochondrial iron movement and consequent iron-dependent mitochondrial ROS formation is a promising strategy to intervene against APAP hepatotoxicity in a clinical setting.
Collapse
Affiliation(s)
- Jiangting Hu
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anna-Liisa Nieminen
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Zhi Zhong
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
3
|
Jaeschke H, Ramachandran A. Central Mechanisms of Acetaminophen Hepatotoxicity: Mitochondrial Dysfunction by Protein Adducts and Oxidant Stress. Drug Metab Dispos 2024; 52:712-721. [PMID: 37567742 PMCID: PMC11257690 DOI: 10.1124/dmd.123.001279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Acetaminophen (APAP) is an analgesic and antipyretic drug used worldwide, which is safe at therapeutic doses. However, an overdose can induce liver injury and even liver failure. Mechanistic studies in mice beginning with the seminal papers published by B.B. Brodie's group in the 1970s have resulted in important insight into the pathophysiology. Although the metabolic activation of APAP with generation of a reactive metabolite, glutathione depletion, and protein adduct formation are critical initiating events, more recently, mitochondria have come into focus as an important target and decision point of cell death. This review provides a comprehensive overview of the induction of mitochondrial superoxide and peroxynitrite formation and its propagation through a mitogen-activated protein kinase cascade, the mitochondrial permeability transition pore opening caused by iron-catalyzed protein nitration, and the mitochondria-dependent nuclear DNA fragmentation. In addition, the role of adaptive mechanisms that can modulate the pathophysiology, including autophagy, mitophagy, nuclear erythroid 2 p45-related factor 2 activation, and mitochondrial biogenesis, are discussed. Importantly, it is outlined how the mechanisms elucidated in mice translate to human hepatocytes and APAP overdose patients, and how this mechanistic insight explains the mechanism of action of the clinically approved antidote N-acetylcysteine and led to the recent discovery of a novel compound, fomepizole, which is currently under clinical development. SIGNIFICANCE STATEMENT: Acetaminophen (APAP)-induced liver injury is the most frequent cause of acute liver failure in western countries. Extensive mechanistic research over the last several decades has revealed a central role of mitochondria in the pathophysiology of APAP hepatotoxicity. This review article provides a comprehensive discussion of a) mitochondrial protein adducts and oxidative/nitrosative stress, b) mitochondria-regulated nuclear DNA fragmentation, c) adaptive mechanisms to APAP-induced cellular stress, d) translation of cell death mechanisms to overdose patients, and e) mechanism-based antidotes against APAP-induced liver injury.
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
4
|
Yin X, Ni G, Zhang X, Fu S, Li H, Gao Z. Tyrosine nitration of glucagon impairs its function: Extending the role of heme in T2D pathogenesis. J Inorg Biochem 2024; 255:112519. [PMID: 38507994 DOI: 10.1016/j.jinorgbio.2024.112519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
New studies raise the possibility that the higher glucagon (GCG) level present in type 2 diabetes (T2D) is a compensatory mechanism to enhance β-cell function, rather than induce dysregulated glucose homeostasis, due to an important role for GCG that acts directly within the pancreas on insulin secretion by intra-islet GCG signaling. However, in states of poorly controlled T2D, pancreatic α cell mass increases (overproduced GCG) in response to insufficient insulin secretion, indicating decreased local GCG activity. The reason for this decrease is not clear. Recent evidence has uncovered a new role of heme in cellular signal transduction, and its mechanism involves reversible binding of heme to proteins. Considering that protein tyrosine nitration in diabetic islets increases and glucose-stimulated insulin secretion (GSIS) decreases, we speculated that heme modulates GSIS by transient interaction with GCG and catalyzing its tyrosine nitration, and the tyrosine nitration may impair GCG activity, leading to loss of intra-islet GCG signaling and markedly impaired insulin secretion. Data presented here elucidate a novel role for heme in disrupting local GCG signaling in diabetes. Heme bound to GCG and induced GCG tyrosine nitration. Two tyrosine residues in GCG were both sensitive to the nitrating species. Further, GCG was also demonstrated to be a preferred target peptide for tyrosine nitration by co-incubation with BSA. Tyrosine nitration impaired GCG stimulated cAMP-dependent signaling in islet β cells and decreased insulin release. Our results provided a new role of heme for impaired GSIS in the pathological process of diabetes.
Collapse
Affiliation(s)
- Xiaoying Yin
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Guoqi Ni
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Xuan Zhang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Shitao Fu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Hailing Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China.
| | - Zhonghong Gao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China.
| |
Collapse
|
5
|
Zeng L, Zhang X, Xia M, Ye H, Li H, Gao Z. Heme and Cu 2+-induced vasoactive intestinal peptide (VIP) tyrosine nitration: A possible molecular mechanism for the attenuated anti-inflammatory effect of VIP in inflammatory diseases. Biochimie 2023; 214:176-187. [PMID: 37481062 DOI: 10.1016/j.biochi.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/24/2023]
Abstract
Vasoactive intestinal peptide (VIP) is a neuropeptide that play an important role in immunoregulation and anti-inflammation. Numerous inflammatory/autoimmune disorders are associated with decreased VIP binding ability to receptors and diminished VIP activation of cAMP generation in immune cells. However, the mechanisms linking oxidative/nitrative stress to VIP immune dysfunction remain unknown. It has been reported that the elevated heme or Cu2+ in inflammatory diseases can cause oxidative and nitrative damage to nearby biological targets under high oxidative stress conditions, which affects the structure and activity of linked peptides or proteins. Thus, the VIP down-regulated immune response may be interfered by redox metal catalyzed VIP tyrosine nitration. To explore this, we systematically investigated the possibility of heme or Cu2+ to catalyze VIP tyrosine nitration. The results showed that Tyr10 and Tyr22 of VIP can both be nitrated in heme/H2O2/NO2- system as well as in Cu2+/H2O2/NO2- system. Then, we used synthetic mutant VIPs with tyrosine residues substituted by 3-nitrotyrosine to study the impact of tyrosine nitration on VIP activity in SHSY-5Y cells. Our findings demonstrated that VIP nitration dramatically decreased the content of its α-helix and random coil, suggesting that VIP nitration might reduce its affinity to the receptor. This was further confirmed in the cAMP assay. The results showed that 10 nM of these tyrosine nitrated VIPs could significantly (p < 0.01) decrease cAMP secretion compared to the wild type VIP. Our data reveal that the attenuation of the neuroprotective effect of VIP in inflammation-related diseases might be attributed to metal-catalyzed VIP tyrosine nitration.
Collapse
Affiliation(s)
- Lizhen Zeng
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Xuan Zhang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Mengyang Xia
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Huixian Ye
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China; School of Chemistry and Chemical Engineering, Jinggangshan University, Ji'an, Jiangxi, 343009, PR China.
| | - Hailing Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China.
| | - Zhonghong Gao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan, 430074, PR China.
| |
Collapse
|
6
|
Abstract
Mitochondria are critical organelles responsible for the maintenance of cellular energy homeostasis. Thus, their dysfunction can have severe consequences in cells responsible for energy-intensive metabolic function, such as hepatocytes. Extensive research over the last decades have identified compromised mitochondrial function as a central feature in the pathophysiology of liver injury induced by an acetaminophen (APAP) overdose, the most common cause of acute liver failure in the United States. While hepatocyte mitochondrial oxidative and nitrosative stress coupled with induction of the mitochondrial permeability transition are well recognized after an APAP overdose, recent studies have revealed additional details about the organelle's role in APAP pathophysiology. This concise review highlights these new advances, which establish the central role of the mitochondria in APAP pathophysiology, and places them in the context of earlier information in the literature. Adaptive alterations in mitochondrial morphology as well as the role of cellular iron in mitochondrial dysfunction and the organelle's importance in liver recovery after APAP-induced injury will be discussed.
Collapse
|
7
|
Mukherjee AG, Valsala Gopalakrishnan A. The interplay of arsenic, silymarin, and NF-ĸB pathway in male reproductive toxicity: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114614. [PMID: 36753973 DOI: 10.1016/j.ecoenv.2023.114614] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Arsenic toxicity is one of the most trending reasons for several malfunctions, particularly reproductive toxicity. The exact mechanism of arsenic poisoning is a big question mark. Exposure to arsenic reduces sperm count, impairs fertilization, and causes inflammation and genotoxicity through interfering with autophagy, epigenetics, ROS generation, downregulation of essential protein expression, metabolite changes, and hampering several signaling cascades, particularly by the alteration of NF-ĸB pathway. This work tries to give a clear idea about the different aspects of arsenic resulting in male reproductive complications, often leading to infertility. The first part of this article explains the implications of arsenic poisoning and the crosstalk of the NF-ĸB pathway in male reproductive toxicity. Silymarin is a bioactive compound that exerts anti-cancer and anti-inflammatory properties and has demonstrated hopeful outcomes in several cancers, including colon cancer, breast cancer, and skin cancer, by downregulating the hyperactive NF-ĸB pathway. The next half of this article thus sheds light on silymarin's therapeutic potential in inhibiting the NF-ĸB signaling cascade, thus offering protection against arsenic-induced male reproductive toxicity.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| |
Collapse
|
8
|
Brasil FB, de Almeida FJS, Luckachaki MD, Dall'Oglio EL, de Oliveira MR. The isothiocyanate sulforaphane prevents mitochondrial impairment and neuroinflammation in the human dopaminergic SH-SY5Y and in the mouse microglial BV2 cells: role for heme oxygenase-1. Metab Brain Dis 2023; 38:419-435. [PMID: 35469083 DOI: 10.1007/s11011-022-00990-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/18/2022] [Indexed: 01/25/2023]
Abstract
Sulforaphane (SFN) promotes protective effects in different cell types. Nonetheless, it remains to be clarified by which mechanism SFN exerts benefits in mammalian cells. Mitochondria are a major source of adenosine triphosphate (ATP) and reactive species in nucleated cells. Mitochondrial impairment result in cellular redox biology disruption, bioenergetic status collapse, and inflammation. Evidence suggest that mitochondrial dysfunction plays a role in neurological disorders. Since a cure was not discovered yet to some of these diseases, investigating strategies to promote mitochondrial protection is pharmacologically relevant and may improve life quality of patients suffering from these maladies. Natural molecules, such as SFN, are potent inducers of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and, consequently, stimulate the expression of genes whose products, such as heme oxygenase-1 (HO-1), induce cytoprotective actions in mammalian tissues. In this work, we investigated whether SFN (5 µM) would be capable to prevent the dysfunctions caused by chlorpyrifos (CPF) on the human dopaminergic SH-SY5Y cells. Moreover, we examined the effects of a pretreatment with SFN at the same concentration on the mouse microglial BV2 cells stimulated by lipopolysaccharide (LPS) in an experimental model of neuroinflammation. SFN prevented the mitochondrial impairment and the neuroinflammation caused by the chemical stressors in both cell types. Inhibition of heme oxygenase-1 (HO-1) suppressed the mitochondrial protection and anti-inflammatory action afforded by SFN in this experimental model. Overall, SFN promoted cytoprotection by a mechanism dependent on the HO-1 enzyme in the SH-SY5Y and BV2 cells.
Collapse
Affiliation(s)
- Flávia Bittencourt Brasil
- Departamento de Ciências da Natureza, Campus Universitário de Rio das Ostras-Universidade Federal Fluminense (UFF), Rio de Janeiro, Brazil
| | - Fhelipe Jolner Souza de Almeida
- Programa de Pós-Graduação Em Ciências da Saúde (PPGCS), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
| | - Matheus Dargesso Luckachaki
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
| | - Evandro Luiz Dall'Oglio
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
| | - Marcos Roberto de Oliveira
- Grupo de Estudos Em Neuroquímica E Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil.
| |
Collapse
|
9
|
Yang B, Li J, Li Y, Zhang M, Zhu J, Zhou T, Deng J. Electrochemical post-treatment of bimetallic-ICP/RGO precursor for Z-scheme CuOx·Ag2O/RGO hetero-structure with catalytic activity enhancement for visible-light-driven photo-Fenton degradation of tetracycline. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2022.121741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
10
|
Adelusi OB, Ramachandran A, Lemasters JJ, Jaeschke H. The role of Iron in lipid peroxidation and protein nitration during acetaminophen-induced liver injury in mice. Toxicol Appl Pharmacol 2022; 445:116043. [PMID: 35513057 PMCID: PMC9843742 DOI: 10.1016/j.taap.2022.116043] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 01/19/2023]
Abstract
Acetaminophen (APAP) hepatotoxicity, a leading cause of acute liver failure in western countries, is characterized by mitochondrial superoxide and peroxynitrite formation. However, the role of iron, especially as facilitator of lipid peroxidation (LPO), has been controversial. Our aim was to determine the mechanism by which iron promotes cell death in this context. Fasted male C57BL/6J mice were treated with the iron chelator deferoxamine, minocycline (inhibitor of the mitochondrial calcium uniporter) or vehicle 1 h before 300 mg/kg APAP. Deferoxamine and minocycline significantly attenuated APAP-induced elevations in serum alanine amino transferase levels and hepatic necrosis at 6 h. This protection correlated with reduced 3-nitro-tyrosine protein adducts; LPO (malondialdehyde, 4-hydroxynonenal) was not detected. Activation of c-jun N-terminal kinase (JNK) was not affected but mitochondrial release of intermembrane proteins was reduced suggesting that the effect of iron was at the level of mitochondria. Co-treatment of APAP with FeSO4 exacerbated liver injury and protein nitration and triggered significant LPO; all effects were reversed by deferoxamine. Thus, after APAP overdose, iron imported into mitochondria facilitates protein nitration by peroxynitrite triggering mitochondrial dysfunction and cell death. Under these conditions, endogenous defense mechanisms largely prevent LPO. However, after iron overload, protein nitration and LPO contribute to APAP hepatotoxicity.
Collapse
Affiliation(s)
- Olamide B Adelusi
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - John J Lemasters
- Center for Cell Death, Injury & Regeneration, Departments of Drug Discovery & Biomedical Sciences and Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
11
|
Kosmachevskaya OV, Nasybullina EI, Shumaev KB, Novikova NN, Topunov AF. Protective Effect of Dinitrosyl Iron Complexes Bound with Hemoglobin on Oxidative Modification by Peroxynitrite. Int J Mol Sci 2021; 22:13649. [PMID: 34948445 PMCID: PMC8703631 DOI: 10.3390/ijms222413649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 12/21/2022] Open
Abstract
Dinitrosyl iron complexes (DNICs) are a physiological form of nitric oxide (•NO) in an organism. They are able not only to deposit and transport •NO, but are also to act as antioxidant and antiradical agents. However, the mechanics of hemoglobin-bound DNICs (Hb-DNICs) protecting Hb against peroxynitrite-caused, mediated oxidative modification have not yet been scrutinized. Through EPR spectroscopy we show that Hb-DNICs are destroyed under the peroxynitrite action in a dose-dependent manner. At the same time, DNICs inhibit the oxidation of tryptophan and tyrosine residues and formation of carbonyl derivatives. They also prevent the formation of covalent crosslinks between Hb subunits and degradation of a heme group. These effects can arise from the oxoferryl heme form being reduced, and they can be connected with the ability of DNICs to directly intercept peroxynitrite and free radicals, which emerge due to its homolysis. These data show that DNICs may ensure protection from myocardial ischemia.
Collapse
Affiliation(s)
- Olga V. Kosmachevskaya
- Research Center of Biotechnology of the Russian Academy of Sciences, Bach Institute of Biochemistry, 119071 Moscow, Russia; (O.V.K.); (E.I.N.); (K.B.S.)
| | - Elvira I. Nasybullina
- Research Center of Biotechnology of the Russian Academy of Sciences, Bach Institute of Biochemistry, 119071 Moscow, Russia; (O.V.K.); (E.I.N.); (K.B.S.)
| | - Konstantin B. Shumaev
- Research Center of Biotechnology of the Russian Academy of Sciences, Bach Institute of Biochemistry, 119071 Moscow, Russia; (O.V.K.); (E.I.N.); (K.B.S.)
| | | | - Alexey F. Topunov
- Research Center of Biotechnology of the Russian Academy of Sciences, Bach Institute of Biochemistry, 119071 Moscow, Russia; (O.V.K.); (E.I.N.); (K.B.S.)
| |
Collapse
|
12
|
Jaeschke H, Adelusi OB, Akakpo JY, Nguyen NT, Sanchez-Guerrero G, Umbaugh DS, Ding WX, Ramachandran A. Recommendations for the use of the acetaminophen hepatotoxicity model for mechanistic studies and how to avoid common pitfalls. Acta Pharm Sin B 2021; 11:3740-3755. [PMID: 35024303 PMCID: PMC8727921 DOI: 10.1016/j.apsb.2021.09.023] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/22/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (APAP) is a widely used analgesic and antipyretic drug, which is safe at therapeutic doses but can cause severe liver injury and even liver failure after overdoses. The mouse model of APAP hepatotoxicity recapitulates closely the human pathophysiology. As a result, this clinically relevant model is frequently used to study mechanisms of drug-induced liver injury and even more so to test potential therapeutic interventions. However, the complexity of the model requires a thorough understanding of the pathophysiology to obtain valid results and mechanistic information that is translatable to the clinic. However, many studies using this model are flawed, which jeopardizes the scientific and clinical relevance. The purpose of this review is to provide a framework of the model where mechanistically sound and clinically relevant data can be obtained. The discussion provides insight into the injury mechanisms and how to study it including the critical roles of drug metabolism, mitochondrial dysfunction, necrotic cell death, autophagy and the sterile inflammatory response. In addition, the most frequently made mistakes when using this model are discussed. Thus, considering these recommendations when studying APAP hepatotoxicity will facilitate the discovery of more clinically relevant interventions.
Collapse
Key Words
- AIF, apoptosis-inducing factor
- AMPK, AMP-activated protein kinase
- APAP, acetaminophen
- ARE, antioxidant response element
- ATG, autophagy-related genes
- Acetaminophen hepatotoxicity
- Apoptosis
- Autophagy
- BSO, buthionine sulfoximine
- CAD, caspase-activated DNase
- CYP, cytochrome P450 enzymes
- DAMPs, damage-associated molecular patterns
- DMSO, dimethylsulfoxide
- Drug metabolism
- EndoG, endonuclease G
- FSP1, ferroptosis suppressing protein 1
- Ferroptosis
- GPX4, glutathione peroxidase 4
- GSH, glutathione
- GSSG, glutathione disulfide
- Gclc, glutamate–cysteine ligase catalytic subunit
- Gclm, glutamate–cysteine ligase modifier subunit
- HMGB1, high mobility group box protein 1
- HNE, 4-hydroxynonenal
- Innate immunity
- JNK, c-jun N-terminal kinase
- KEAP1, Kelch-like ECH-associated protein 1
- LAMP, lysosomal-associated membrane protein
- LC3, light chain 3
- LOOH, lipid hydroperoxides
- LPO, lipid peroxidation
- MAP kinase, mitogen activated protein kinase
- MCP-1, monocyte chemoattractant protein-1
- MDA, malondialdehyde
- MPT, mitochondrial permeability transition
- Mitochondria
- MnSOD, manganese superoxide dismutase
- NAC, N-acetylcysteine
- NAPQI, N-acetyl-p-benzoquinone imine
- NF-κB, nuclear factor κB
- NQO1, NAD(P)H:quinone oxidoreductase 1
- NRF2
- NRF2, nuclear factor erythroid 2-related factor 2
- PUFAs, polyunsaturated fatty acids
- ROS, reactive oxygen species
- SMAC/DIABLO, second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low pI
- TLR, toll like receptor
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- UGT, UDP-glucuronosyltransferases
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Olamide B. Adelusi
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jephte Y. Akakpo
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nga T. Nguyen
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Giselle Sanchez-Guerrero
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - David S. Umbaugh
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
13
|
Kosmachevskaya OV, Nasybullina EI, Shumaev KB, Chumikina LV, Arabova LI, Yaglova NV, Obernikhin SS, Topunov AF. Dinitrosyl Iron Complexes with Glutathione Ligands Intercept Peroxynitrite and Protect Hemoglobin from Oxidative Modification. APPL BIOCHEM MICRO+ 2021. [DOI: 10.1134/s0003683821040098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
14
|
3-Nitrotyrosine and related derivatives in proteins: precursors, radical intermediates and impact in function. Essays Biochem 2020; 64:111-133. [PMID: 32016371 DOI: 10.1042/ebc20190052] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/30/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022]
Abstract
Oxidative post-translational modification of proteins by molecular oxygen (O2)- and nitric oxide (•NO)-derived reactive species is a usual process that occurs in mammalian tissues under both physiological and pathological conditions and can exert either regulatory or cytotoxic effects. Although the side chain of several amino acids is prone to experience oxidative modifications, tyrosine residues are one of the preferred targets of one-electron oxidants, given the ability of their phenolic side chain to undergo reversible one-electron oxidation to the relatively stable tyrosyl radical. Naturally occurring as reversible catalytic intermediates at the active site of a variety of enzymes, tyrosyl radicals can also lead to the formation of several stable oxidative products through radical-radical reactions, as is the case of 3-nitrotyrosine (NO2Tyr). The formation of NO2Tyr mainly occurs through the fast reaction between the tyrosyl radical and nitrogen dioxide (•NO2). One of the key endogenous nitrating agents is peroxynitrite (ONOO-), the product of the reaction of superoxide radical (O2•-) with •NO, but ONOO--independent mechanisms of nitration have been also disclosed. This chemical modification notably affects the physicochemical properties of tyrosine residues and because of this, it can have a remarkable impact on protein structure and function, both in vitro and in vivo. Although low amounts of NO2Tyr are detected under basal conditions, significantly increased levels are found at pathological states related with an overproduction of reactive species, such as cardiovascular and neurodegenerative diseases, inflammation and aging. While NO2Tyr is a well-established stable oxidative stress biomarker and a good predictor of disease progression, its role as a pathogenic mediator has been laboriously defined for just a small number of nitrated proteins and awaits further studies.
Collapse
|
15
|
H M, S H, N QI, R M, A M, R S, L S S, K B, P B, H D, P N N, M M, Y N. Surface refined Au Quercetin nanoconjugate stimulates dermal cell migration: possible implication in wound healing. RSC Adv 2020; 10:37683-37694. [PMID: 35515178 PMCID: PMC9057138 DOI: 10.1039/d0ra06690g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/16/2020] [Indexed: 01/13/2023] Open
Abstract
Refining nutraceutical conjugated metal nanoparticles (NPs) and understanding their interactions with the cellular micro-environment is necessary for their application in nanomedicine. In the present experiment, we studied the effect of quercetin functionalized gold nanoparticles (AuQurNP) on skin fibroblast and keratinocyte cell migration. Spherical shaped AuQurNPs of 47 nm in size were formed due to the interaction of hydroxyl and carbonyl groups of quercetin with Au atoms as revealed by incremental algorithm-based analysis. AuQurNP containing up to 5 μg l−1 of Au with quercetin (5.2 ± 1.6 ng ml−1) was least toxic to fibroblasts. AuQurNP effectively reduced the generation of intracellular ROS (up to 63%) through free-radical scavenging activity. AuQurNP also enhanced the rate of migration of fibroblasts (24 h) and keratinocytes (20 h) in artificially created wounds. The rate of migration of the cells towards the wound edge was in the order of AuQurNP > control > quercetin > AuNP. AuQurNP also significantly increased the expression of TGFβ1 protein, thereby inducing the downstream SMAD complex (SMAD 2–4). Downregulation of the inhibitory protein SMAD 7 by AuQurNP helped in the nuclear translocation of SMADs 3 and 4. Collectively, the present in vitro study demonstrates the action of AuQurNP on the SMAD family and the interconnected molecular mechanism leading to the cell migration process. AuQuercetin nano conjugates enhances cell migration via TGFβ1.![]()
Collapse
Affiliation(s)
- Madhyastha H
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki Miyazaki 889 1692 Japan
| | - Halder S
- School of Biosciences and Technology, Vellore Institute of Technology Vellore 632014 Tamilnadu India
| | - Queen Intan N
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki Miyazaki 889 1692 Japan
| | - Madhyastha R
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki Miyazaki 889 1692 Japan
| | - Mohanapriya A
- School of Biosciences and Technology, Vellore Institute of Technology Vellore 632014 Tamilnadu India
| | - Sudhakaran R
- School of Biosciences and Technology, Vellore Institute of Technology Vellore 632014 Tamilnadu India
| | - Sajitha L S
- School of Biosciences and Technology, Vellore Institute of Technology Vellore 632014 Tamilnadu India
| | - Banerjee K
- School of Biosciences and Technology, Vellore Institute of Technology Vellore 632014 Tamilnadu India
| | - Bethasiwi P
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki Miyazaki 889 1692 Japan
| | - Daima H
- Amity Center for Nanobiotechnology and Nanomedicine, Amity Institute of Biotechnology, Amity University Rajasthan Jaipur 303002 Rajasthan India
| | - Navya P N
- Department of Biotechnology, Bannari Amman Institute of Technology Sathyamangalam Erode 638401 Tamilnadu India
| | - Maruyama M
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki Miyazaki 889 1692 Japan
| | - Nakajima Y
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki Miyazaki 889 1692 Japan
| |
Collapse
|
16
|
Palma JM, Mateos RM, López-Jaramillo J, Rodríguez-Ruiz M, González-Gordo S, Lechuga-Sancho AM, Corpas FJ. Plant catalases as NO and H 2S targets. Redox Biol 2020; 34:101525. [PMID: 32505768 PMCID: PMC7276441 DOI: 10.1016/j.redox.2020.101525] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022] Open
Abstract
Catalase is a powerful antioxidant metalloenzyme located in peroxisomes which also plays a central role in signaling processes under physiological and adverse situations. Whereas animals contain a single catalase gene, in plants this enzyme is encoded by a multigene family providing multiple isoenzymes whose number varies depending on the species, and their expression is regulated according to their tissue/organ distribution and the environmental conditions. This enzyme can be modulated by reactive oxygen and nitrogen species (ROS/RNS) as well as by hydrogen sulfide (H2S). Catalase is the major protein undergoing Tyr-nitration [post-translational modification (PTM) promoted by RNS] during fruit ripening, but the enzyme from diverse sources is also susceptible to undergo other activity-modifying PTMs. Data on S-nitrosation and persulfidation of catalase from different plant origins are given and compared here with results from obese children where S-nitrosation of catalase occurs. The cysteine residues prone to be S-nitrosated in catalase from plants and from bovine liver have been identified. These evidences assign to peroxisomes a crucial statement in the signaling crossroads among relevant molecules (NO and H2S), since catalase is allocated in these organelles. This review depicts a scenario where the regulation of catalase through PTMs, especially S-nitrosation and persulfidation, is highlighted.
Collapse
Affiliation(s)
- José M Palma
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Dept. Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Granada, Spain.
| | - Rosa M Mateos
- Imflammation, Nutrition, Metabolism and Oxidative Stress Study Group (INMOX), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cádiz, Spain; Area of Biochemistry and Molecular Biology, Department of Biomedicine, Biotechnology and Public Health, University of Cádiz, Cádiz, Spain
| | | | - Marta Rodríguez-Ruiz
- Laboratório de Fisiologia do Desenvolvimiento Vegetal; Instituto de Biociências-Universidad de São Paulo; Cidade Universitária-São Paulo-SP, Brazil
| | - Salvador González-Gordo
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Dept. Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Granada, Spain
| | - Alfonso M Lechuga-Sancho
- Imflammation, Nutrition, Metabolism and Oxidative Stress Study Group (INMOX), Biomedical Research and Innovation Institute of Cádiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cádiz, Spain; Department of Child and Mother Health and Radiology, Medical School, University of Cádiz, Cádiz, Spain
| | - Francisco J Corpas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Dept. Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Granada, Spain
| |
Collapse
|
17
|
Fang X, Gao W, Yang Z, Gao Z, Li H. Dual Anti-/Prooxidant Behaviors of Flavonoids Pertaining to Cu(II)-Catalyzed Tyrosine Nitration of the Insulin Receptor Kinase Domain in an Antidiabetic Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6202-6211. [PMID: 32395994 DOI: 10.1021/acs.jafc.0c01676] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Flavonoid, as a potent antioxidant, exerts many beneficial effects in type 2 diabetes, whereas the prooxidative property may be also important in vivo if copper is involved. Here, we chose an insulin receptor kinase domain fragment (KK-1, residues 1126-1165), containing the A-loop of the receptor as well as three key autophosphorylation sites (Tyr1158, Tyr1162, and Tyr1163) associated with receptor signal transduction to investigate the roles and the structure-activity relationship of three antidiabetic flavonoids (kaempferol, luteolin, and apigenin) and two others with a similar structure (diosmetin and genistein), on modulation of Cu(II)-mediated tyrosine nitration and the corresponding effect on its functional phosphorylation in the Cu2+/H2O2/NO2- system. We found that both properties of flavonoid played roles on inhibition of Cu(II)-mediated protein nitration in the H2O2/NO2- system: (1) on the one hand, flavonoid scavenged free radicals as antioxidants, inhibited tyrosine nitration, and thus inhibited the reduction of tyrosine phosphorylation caused by tyrosine nitration; and (2) on the other hand, flavonoid promoted •OH production as a prooxidant, which increased 3,3'-dityrosine formation. The formation of 3,3'-dityrosine decreased Cu2+-induced tyrosine nitration and thus interfered with its phosphorylation. This study confirms that the weight relationship between antioxidation and prooxidation of a flavonoid needs to be studied clearly before nutritional and medical applications.
Collapse
Affiliation(s)
- Xu Fang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, P. R. China
| | - Wanxia Gao
- School of Basic Medical Science, Hubei University of Science and Technology, Xianning 437000, P. R. China
| | - Zhen Yang
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77004, United States
| | - Zhonghong Gao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, P. R. China
| | - Hailing Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, P. R. China
| |
Collapse
|
18
|
Rodríguez-Ruiz M, González-Gordo S, Cañas A, Campos MJ, Paradela A, Corpas FJ, Palma JM. Sweet Pepper ( Capsicum annuum L.) Fruits Contain an Atypical Peroxisomal Catalase That is Modulated by Reactive Oxygen and Nitrogen Species. Antioxidants (Basel) 2019; 8:E374. [PMID: 31487955 PMCID: PMC6769641 DOI: 10.3390/antiox8090374] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/20/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
Abstract
During the ripening of sweet pepper (Capsicum annuum L.) fruits, in a genetically controlled scenario, enormous metabolic changes occur that affect the physiology of most cell compartments. Peroxisomal catalase gene expression decreases after pepper fruit ripening, while the enzyme is also susceptible to undergo post-translational modifications (nitration, S-nitrosation, and oxidation) promoted by reactive oxygen and nitrogen species (ROS/RNS). Unlike most plant catalases, the pepper fruit enzyme acts as a homodimer, with an atypical native molecular mass of 125 to 135 kDa and an isoelectric point of 7.4, which is higher than that of most plant catalases. These data suggest that ROS/RNS could be essential to modulate the role of catalase in maintaining basic cellular peroxisomal functions during pepper fruit ripening when nitro-oxidative stress occurs. Using catalase from bovine liver as a model and biotin-switch labeling, in-gel trypsin digestion, and nanoliquid chromatography coupled with mass spectrometry, it was found that Cys377 from the bovine enzyme could potentially undergo S-nitrosation. To our knowledge, this is the first report of a cysteine residue from catalase that can be post-translationally modified by S-nitrosation, which makes it especially important to find the target points where the enzyme can be modulated under either physiological or adverse conditions.
Collapse
Affiliation(s)
- Marta Rodríguez-Ruiz
- Group Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| | - Salvador González-Gordo
- Group Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| | - Amanda Cañas
- Group Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| | - María Jesús Campos
- Group Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| | - Alberto Paradela
- Proteomics Core Facility, Centro Nacional de Biotecnología, CSIC, 28049 Madrid, Spain.
| | - Francisco J Corpas
- Group Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| | - José M Palma
- Group Antioxidant, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain.
| |
Collapse
|
19
|
Structure effect of water-soluble iron porphyrins on catalyzing protein tyrosine nitration in the presence of nitrite and hydrogen peroxide. Nitric Oxide 2019; 91:42-51. [PMID: 31351146 DOI: 10.1016/j.niox.2019.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022]
Abstract
Water-soluble iron porphyrins, such as FeTPPS (5,10,15,20-tetrakis (4-sulfonatophenyl) porphyrinato iron (III)), FeTMPyP (5,10,15,20-tetrakis (N-methyl-4'-pyridyl) porphyrinato iron (III) chloride) and FeTBAP (5,10,15,20-tetrakis (4-benzoic acid) porphyrinato iron (III)), are highly active catalysts for peroxynitrite decomposition and thereby have been suggested as therapeutic agent for inflammatory diseases that implicate the involvement of nitrotyrosine formation. Here, we systemically investigated catalytic properties of FeTPPS, FeTMPyP and FeTBAP on protein nitration in the presence of hydrogen peroxide and nitrite. We showed that FeTPPS, FeTBAP and FeTMPyP all exhibited higher peroxidase activity in compared with hemin. As to protein nitration, the catalytic effect of FeTPPS and FeTBAP are effective in the presence of hydrogen peroxide and nitrite, while negligible BSA nitration was observed in the case of FeTMPyP. Moreover, the underlying mechanism of the oxidation of FeTPPS, FeTBAP and FeTMPyP was further studied. Collectively, our results suggest that, compound I and II species are involved in as the key intermediates in FeTMPyP/H2O2 system as similar as those in FeTPPS/H2O2 and FeTBAP/H2O2 system. As compared to weak antioxidants, TPPS and TBAP, however, TMPyP scavenges oxo-Fe (IV) intermediates of FeTMPyP at a faster rate by significant self-degradation; results in the shortest lifetimes of OFeIV-TMPyP and the lowest catalytic activity on oxidizing tyrosine and nitrite; and therefore, attributes to inactivation of FeTMPyP in protein nitration. In addition, association of FeTMPyP to BSA was found weak, while strong binding of FeTPPS and FeTBAP were observed. The weak binding keeps away of target residue of BSA from the center of FeTMPyP where the RNS is generated, which might be attributed as additional factors to the inactivation of FeTMPyP in protein nitration.
Collapse
|
20
|
Nitroso-Oxidative Stress, Acute Phase Response, and Cytogenetic Damage in Wistar Rats Treated with Adrenaline. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1805354. [PMID: 30584458 PMCID: PMC6280229 DOI: 10.1155/2018/1805354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/19/2018] [Accepted: 09/12/2018] [Indexed: 12/14/2022]
Abstract
This study is aimed at analysing biochemical and genetic endpoints of toxic effects after administration of adrenaline. For this purpose, the study was carried out on Wistar rats and three doses of adrenaline were used: 0.75 mg/kg, 1.5 mg/kg, and 3 mg/kg body weight. To achieve these aims, we investigated the effects of adrenaline on catalase (CAT), Cu, Zn-superoxide dismutase (SOD), malondialdehyde (MDA), nitrite (NO2−), carbonyl groups (PCC), and nitrotyrosine (3-NT). Total activity of lactate dehydrogenase (LDH), its relative distribution (LDH1–LDH5) activity, level of acute phase proteins (APPs), and genotoxic effect were also evaluated. The obtained results revealed that all doses of adrenaline induced a significant rise in CAT activity, MDA level, PCC, NO2−, and 3-NT and a significant decrease in SOD activity compared to control. Adrenaline exerted an increase in total activity of LDH, LDH1, and LDH2 isoenzymes. Further study showed that adrenaline significantly decreased serum albumin level and albumin-globulin ratio, while the level of APPs (α1-acid glycoprotein and haptoglobulin) is increased. The micronucleus test revealed a genotoxic effect of adrenaline at higher concentrations (1.5 mg/kg and 3 mg/kg body weight) compared to untreated rats. It can be concluded that adrenaline exerts oxidative and nitrative stress in rats, increased damage to lipids and proteins, and damage of cardiomyocytes and cytogenetic damage. Obtained results may contribute to better understanding of the toxicity of adrenaline with aims to preventing its harmful effects.
Collapse
|
21
|
Tyrosine residues of bovine serum albumin play an important role in protecting SH-SY5Y cells against heme/H2O2/NO2−-induced damage. Mol Cell Biochem 2018; 454:57-66. [DOI: 10.1007/s11010-018-3452-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/05/2018] [Indexed: 02/08/2023]
|
22
|
Zhang P, Ma L, Yang Z, Li H, Gao Z. 5,10,15,20-Tetrakis(4-sulfonatophenyl)porphyrinato iron(III) chloride (FeTPPS), a peroxynitrite decomposition catalyst, catalyzes protein tyrosine nitration in the presence of hydrogen peroxide and nitrite. J Inorg Biochem 2018. [DOI: 10.1016/j.jinorgbio.2018.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
23
|
Bartesaghi S, Radi R. Fundamentals on the biochemistry of peroxynitrite and protein tyrosine nitration. Redox Biol 2018; 14:618-625. [PMID: 29154193 PMCID: PMC5694970 DOI: 10.1016/j.redox.2017.09.009] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/06/2017] [Accepted: 09/15/2017] [Indexed: 12/26/2022] Open
Abstract
In this review we provide an analysis of the biochemistry of peroxynitrite and tyrosine nitration. Peroxynitrite is the product of the diffusion-controlled reaction between superoxide (O2•-) and nitric oxide (•NO). This process is in competition with the enzymatic dismutation of O2•- and the diffusion of •NO across cells and tissues and its reaction with molecular targets (e.g. guanylate cyclase). Understanding the kinetics and compartmentalization of the O2•- / •NO interplay is critical to rationalize the shift of •NO from a physiological mediator to a cytotoxic intermediate. Once formed, peroxynitrite (ONOO- and ONOOH; pKa = 6,8) behaves as a strong one and two-electron oxidant towards a series of biomolecules including transition metal centers and thiols. In addition, peroxynitrite anion can secondarily evolve to secondary radicals either via its fast reaction with CO2 or through proton-catalyzed homolysis. Thus, peroxynitrite can participate in direct (bimolecular) and indirect (through secondary radical intermediates) oxidation reactions; through these processes peroxynitrite can participate as cytotoxic effector molecule against invading pathogens and/or as an endogenous pathogenic mediator. Peroxynitrite can cause protein tyrosine nitration in vitro and in vivo. Indeed, tyrosine nitration is a hallmark of the reactions of •NO-derived oxidants in cells and tissues and serves as a biomarker of oxidative damage. Protein tyrosine nitration can mediate changes in protein structure and function that affect cell homeostasis. Tyrosine nitration in biological systems is a free radical process that can be promoted either by peroxynitrite-derived radicals or by other related •NO-dependent oxidative processes. Recently, mechanisms responsible of tyrosine nitration in hydrophobic biostructures such as membranes and lipoproteins have been assessed and involve the parallel occurrence and connection with lipid peroxidation. Experimental strategies to reveal the proximal oxidizing mechanism during tyrosine nitration in given pathophysiologically-relevant conditions include mapping and identification of the tyrosine nitration sites in specific proteins.
Collapse
Affiliation(s)
- Silvina Bartesaghi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. General Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. General Flores 2125, Montevideo 11800, Uruguay.
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. General Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. General Flores 2125, Montevideo 11800, Uruguay.
| |
Collapse
|
24
|
Ferreira CM, Oliveira MP, Paes MC, Oliveira MF. Modulation of mitochondrial metabolism as a biochemical trait in blood feeding organisms: the redox vampire hypothesis redux. Cell Biol Int 2018; 42:683-700. [PMID: 29384241 DOI: 10.1002/cbin.10945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/27/2018] [Indexed: 12/31/2022]
Abstract
Hematophagous organisms undergo remarkable metabolic changes during the blood digestion process, increasing fermentative glucose metabolism, and reducing respiratory rates, both consequence of functional mitochondrial remodeling. Here, we review the pathways involved in energy metabolism and mitochondrial functionality in a comparative framework across different hematophagous species, and consider how these processes regulate redox homeostasis during blood digestion. The trend across distinct species indicate that a switch in energy metabolism might represent an important defensive mechanism to avoid the potential harmful interaction of oxidants generated from aerobic energy metabolism with products derived from blood digestion. Indeed, in insect vectors, blood feeding transiently reduces respiratory rates and oxidant production, irrespective of tissue and insect model. On the other hand, a different scenario is observed in several unrelated parasite species when exposed to blood digestion products, as respiratory rates reduce and mitochondrial oxidant production increase. The emerging picture indicates that re-wiring of energy metabolism, through reduced mitochondrial function, culminates in improved tolerance to redox insults and seems to represent a key step for hematophagous organisms to cope with the overwhelming and potentially toxic blood meal.
Collapse
Affiliation(s)
- Caroline M Ferreira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-590, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, 21941-590, Brazil
| | - Matheus P Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-590, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, 21941-590, Brazil.,Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Marcia C Paes
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, 21941-590, Brazil.,Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Marcus F Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-590, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, 21941-590, Brazil
| |
Collapse
|
25
|
Ferrer-Sueta G, Campolo N, Trujillo M, Bartesaghi S, Carballal S, Romero N, Alvarez B, Radi R. Biochemistry of Peroxynitrite and Protein Tyrosine Nitration. Chem Rev 2018; 118:1338-1408. [DOI: 10.1021/acs.chemrev.7b00568] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Gerardo Ferrer-Sueta
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Nicolás Campolo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Silvina Bartesaghi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Sebastián Carballal
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Romero
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Beatriz Alvarez
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
26
|
Sharma SK, Schaefer AW, Lim H, Matsumura H, Moënne-Loccoz P, Hedman B, Hodgson KO, Solomon EI, Karlin KD. A Six-Coordinate Peroxynitrite Low-Spin Iron(III) Porphyrinate Complex-The Product of the Reaction of Nitrogen Monoxide (·NO (g)) with a Ferric-Superoxide Species. J Am Chem Soc 2017; 139:17421-17430. [PMID: 29091732 PMCID: PMC5783694 DOI: 10.1021/jacs.7b08468] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Peroxynitrite (-OON═O, PN) is a reactive nitrogen species (RNS) which can effect deleterious nitrative or oxidative (bio)chemistry. It may derive from reaction of superoxide anion (O2•-) with nitric oxide (·NO) and has been suggested to form an as-yet unobserved bound heme-iron-PN intermediate in the catalytic cycle of nitric oxide dioxygenase (NOD) enzymes, which facilitate a ·NO homeostatic process, i.e., its oxidation to the nitrate anion. Here, a discrete six-coordinate low-spin porphyrinate-FeIII complex [(PIm)FeIII(-OON═O)] (3) (PIm; a porphyrin moiety with a covalently tethered imidazole axial "base" donor ligand) has been identified and characterized by various spectroscopies (UV-vis, NMR, EPR, XAS, resonance Raman) and DFT calculations, following its formation at -80 °C by addition of ·NO(g) to the heme-superoxo species, [(PIm)FeIII(O2•-)] (2). DFT calculations confirm that 3 is a six-coordinate low-spin species with the PN ligand coordinated to iron via its terminal peroxidic anionic O atom with the overall geometry being in a cis-configuration. Complex 3 thermally transforms to its isomeric low-spin nitrato form [(PIm)FeIII(NO3-)] (4a). While previous (bio)chemical studies show that phenolic substrates undergo nitration in the presence of PN or PN-metal complexes, in the present system, addition of 2,4-di-tert-butylphenol (2,4DTBP) to complex 3 does not lead to nitrated phenol; the nitrate complex 4a still forms. DFT calculations reveal that the phenolic H atom approaches the terminal PN O atom (farthest from the metal center and ring core), effecting O-O cleavage, giving nitrogen dioxide (·NO2) plus a ferryl compound [(PIm)FeIV═O] (7); this rebounds to give [(PIm)FeIII(NO3-)] (4a).The generation and characterization of the long sought after ferriheme peroxynitrite complex has been accomplished.
Collapse
Affiliation(s)
- Savita K. Sharma
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Andrew W. Schaefer
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Hyeongtaek Lim
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Hirotoshi Matsumura
- Division of Environmental & Biomolecular Systems, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
| | - Pierre Moënne-Loccoz
- Division of Environmental & Biomolecular Systems, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
| | - Britt Hedman
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Keith O. Hodgson
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Edward I. Solomon
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Kenneth D. Karlin
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
27
|
Madhyastha H, Madhyastha R, Nakajima Y, Maruyama M. Deciphering the molecular events during arsenic induced transcription signal cascade activation in cellular milieu. Biometals 2017; 31:7-15. [PMID: 29143154 DOI: 10.1007/s10534-017-0065-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/06/2017] [Indexed: 12/18/2022]
Abstract
Anthropogenic sources of arsenic poses and creates unintentional toxico-pathological concerns to humans in many parts of the world. The understanding of toxicity of this metalloid, which shares properties of both metal and non-metal is principally structured on speciation types and holy grail of toxicity prevention. Visible symptoms of arsenic toxicity include nausea, vomiting, diarrhea and abdominal pain. In this review, we focused on the dermal cell stress caused by trivalent arsenic trioxide and pentavalent arsanilic acid. Deciphering the molecular events involved during arsenic toxicity and signaling cascade interaction is key in arsenicosis prevention. FoxO1 and FoxO2 transcription factors, members of the Forkhead/Fox family, play important roles in this aspect. Like Foxo family proteins, ATM/CHK signaling junction also plays important role in DNA nuclear factor guided cellular development. This review will summarize and discuss current knowledge about the interplay of these pathways in arsenic induced dermal pathogenesis.
Collapse
Affiliation(s)
- Harishkumar Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 8891692, Japan
| | - Radha Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 8891692, Japan
| | - Yuichi Nakajima
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 8891692, Japan
| | - Masugi Maruyama
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 8891692, Japan.
| |
Collapse
|
28
|
Karlsson JOG, Andersson RG, Jynge P. Mangafodipir a Selective Cytoprotectant - with Special Reference to Oxaliplatin and Its Association to Chemotherapy-Induced Peripheral Neuropathy (CIPN). Transl Oncol 2017; 10:641-649. [PMID: 28668762 PMCID: PMC5496205 DOI: 10.1016/j.tranon.2017.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 12/12/2022] Open
Abstract
Oxaliplatin, in combination with 5-fluorouracil plus folinate (or capecitabine), has increased survival substantially in stage III colorectal cancer and prolonged life in stage IV patients, but its use is compromised because of severe toxicity. Chemotherapy-induced peripheral neuropathy (CIPN) is the most problematic dose-limiting toxicity of oxaliplatin. Oncologists included for years calcium and magnesium infusion as part of clinical practice for preventing CIPN. Results from a phase III prospective study published in 2014, however, overturned this practice. No other treatments have been clinically proven to prevent this toxicity. There is a body of evidence that CIPN is caused by cellular oxidative stress. Clinical and preclinical data suggest that the manganese chelate and superoxide dismutase mimetic mangafodipir (MnDPDP) is an efficacious inhibitor of CIPN and other conditions caused by cellular oxidative stress, without interfering negatively with the tumoricidal activity of chemotherapy. MnPLED, the metabolite of MnDPDP, attacks cellular oxidative stress at several critical levels. Firstly, MnPLED catalyzes dismutation of superoxide (O2•−), and secondly, having a tremendous high affinity for iron (and copper), PLED binds and disarms redox active iron/copper, which is involved in several detrimental oxidative steps. A case report from 2009 and a recent feasibility study suggest that MnDPDP may prevent or even cure oxaliplatin-induced CIPN. Preliminary results from a phase II study (PLIANT) suggest efficacy also of calmangafodipir, but these results are according to available data obscured by a surprisingly low number of adverse events and a seemingly lower than expected efficacy of FOLFOX.
Collapse
Affiliation(s)
| | - Rolf Gg Andersson
- Division of Drug Research/Pharmacology, Linköping University, Sweden
| | - Per Jynge
- Division of Drug Research/Pharmacology, Linköping University, Sweden
| |
Collapse
|
29
|
Bartesaghi S, Herrera D, Martinez DM, Petruk A, Demicheli V, Trujillo M, Martí MA, Estrín DA, Radi R. Tyrosine oxidation and nitration in transmembrane peptides is connected to lipid peroxidation. Arch Biochem Biophys 2017; 622:9-25. [PMID: 28412156 DOI: 10.1016/j.abb.2017.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 12/30/2022]
Abstract
Tyrosine nitration is an oxidative post-translational modification that can occur in proteins associated to hydrophobic bio-structures such as membranes and lipoproteins. In this work, we have studied tyrosine nitration in membranes using a model system consisting of phosphatidylcholine liposomes with pre-incorporated tyrosine-containing 23 amino acid transmembrane peptides. Tyrosine residues were located at positions 4, 8 or 12 of the amino terminal, resulting in different depths in the bilayer. Tyrosine nitration was accomplished by exposure to peroxynitrite and a peroxyl radical donor or hemin in the presence of nitrite. In egg yolk phosphatidylcholine liposomes, nitration was highest for the peptide with tyrosine at position 8 and dramatically increased as a function of oxygen levels. Molecular dynamics studies support that the proximity of the tyrosine phenolic ring to the linoleic acid peroxyl radicals contributes to the efficiency of tyrosine oxidation. In turn, α-tocopherol inhibited both lipid peroxidation and tyrosine nitration. The mechanism of tyrosine nitration involves a "connecting reaction" by which lipid peroxyl radicals oxidize tyrosine to tyrosyl radical and was fully recapitulated by computer-assisted kinetic simulations. Altogether, this work underscores unique characteristics of the tyrosine oxidation and nitration process in lipid-rich milieu that is fueled via the lipid peroxidation process.
Collapse
Affiliation(s)
- Silvina Bartesaghi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Departamento de Educación Médica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay.
| | - Daniel Herrera
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Débora M Martinez
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Ariel Petruk
- Departamento de Química Inorgánica, Analítica y Química-Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cuidad Universitaria, Pab 2, C1428EHA, Buenos Aires, Argentina
| | - Verónica Demicheli
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Marcelo A Martí
- Departamento de Química Biológica and IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cuidad Universitaria, Pab 2, C1428EHA, Buenos Aires, Argentina
| | - Darío A Estrín
- Departamento de Química Inorgánica, Analítica y Química-Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cuidad Universitaria, Pab 2, C1428EHA, Buenos Aires, Argentina
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay.
| |
Collapse
|
30
|
Resveratrol and Brain Mitochondria: a Review. Mol Neurobiol 2017; 55:2085-2101. [DOI: 10.1007/s12035-017-0448-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/07/2017] [Indexed: 12/24/2022]
|
31
|
Ye H, Yang Z, Li H, Gao Z. NPY binds with heme to form a NPY–heme complex: enhancing peroxidase activity in free heme and promoting NPY nitration and inactivation. Dalton Trans 2017; 46:10315-10323. [DOI: 10.1039/c7dt01822c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
NPY binding with heme enhances the peroxidase activity of free heme, resulting in the important tyrosine nitration, which will attenuate its bioactivity.
Collapse
Affiliation(s)
- Huixian Ye
- School of Chemistry and Chemical Engineering
- Huazhong University of Science and Technology
- Wuhan 430074
- People's Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica
| | - Zhen Yang
- Department of Chemical and Biomolecular Engineering
- University of Houston
- Houston
- USA
| | - Hailing Li
- School of Chemistry and Chemical Engineering
- Huazhong University of Science and Technology
- Wuhan 430074
- People's Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica
| | - Zhonghong Gao
- School of Chemistry and Chemical Engineering
- Huazhong University of Science and Technology
- Wuhan 430074
- People's Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica
| |
Collapse
|
32
|
Wu T, Wang S, Wang Z, Peng X, Lu Y, Wu Q. A multicopper oxidase contributes to the copper tolerance of Brucella melitensis 16M. FEMS Microbiol Lett 2015; 362:fnv078. [DOI: 10.1093/femsle/fnv078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2015] [Indexed: 12/22/2022] Open
|
33
|
Pereira M, Soares C, Canuto GAB, Tavares MFM, Colli W, Alves MJM. Down regulation of NO signaling in Trypanosoma cruzi upon parasite-extracellular matrix interaction: changes in protein modification by nitrosylation and nitration. PLoS Negl Trop Dis 2015; 9:e0003683. [PMID: 25856423 PMCID: PMC4391712 DOI: 10.1371/journal.pntd.0003683] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/06/2015] [Indexed: 01/18/2023] Open
Abstract
Background Adhesion of the Trypanosoma cruzi trypomastigotes, the causative agent of Chagas' disease in humans, to components of the extracellular matrix (ECM) is an important step in host cell invasion. The signaling events triggered in the parasite upon binding to ECM are less explored and, to our knowledge, there is no data available regarding •NO signaling. Methodology/Principal Findings Trypomastigotes were incubated with ECM for different periods of time. Nitrated and S-nitrosylated proteins were analyzed by Western blotting using anti-nitrotyrosine and S-nitrosyl cysteine antibodies. At 2 h incubation time, a decrease in NO synthase activity, •NO, citrulline, arginine and cGMP concentrations, as well as the protein modifications levels have been observed in the parasite. The modified proteins were enriched by immunoprecipitation with anti-nitrotyrosine antibodies (nitrated proteins) or by the biotin switch method (S-nitrosylated proteins) and identified by MS/MS. The presence of both modifications was confirmed in proteins of interest by immunoblotting or immunoprecipitation. Conclusions/Significance For the first time it was shown that T. cruzi proteins are amenable to modifications by S-nitrosylation and nitration. When T. cruzi trypomastigotes are incubated with the extracellular matrix there is a general down regulation of these reactions, including a decrease in both NOS activity and cGMP concentration. Notwithstanding, some specific proteins, such as enolase or histones had, at least, their nitration levels increased. This suggests that post-translational modifications of T. cruzi proteins are not only a reflex of NOS activity, implying other mechanisms that circumvent a relatively low synthesis of •NO. In conclusion, the extracellular matrix, a cell surrounding layer of macromolecules that have to be trespassed by the parasite in order to be internalized into host cells, contributes to the modification of •NO signaling in the parasite, probably an essential move for the ensuing invasion step. Interaction of Trypanosoma cruzi with the extracellular matrix (ECM) is an essential step in the invasion of mammalian cells. However, the nature of the signaling triggered in the parasite is poorly understood. Herein the key role of nitric oxide in T. cruzi signaling is described, using an ECM preparation, in the absence of host cells. Inhibition of NOS activity, with the expected decrease in •NO production, as well as decrease in cGMP concentration were observed by the incubation of T. cruzi trypomastigotes with ECM. Additionally, lower levels of protein S-nitrosylation and nitration were detected. These post-translational modifications have been analyzed by biotin-switch and protein immunoprecipitation approaches coupled to mass spectrometry. The presence of both modifications was confirmed for specific proteins, as mucin II (S-nitrosylation), histones, enolase and tubulins. To our knowledge, decrease in the •NO signaling pathway upon T. cruzi trypomastigotes adhesion to ECM, affecting both the canonical pathway (•NO-soluble guanylyl cyclase-cGMP) and protein S-nitrosylation and nitration is described for the first time in this parasite.
Collapse
Affiliation(s)
- Milton Pereira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Chrislaine Soares
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Walter Colli
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Maria Julia M. Alves
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|